1
|
Xue C, Chen H, Zhao Y, Yuan D, Fang X, Ding M, Qu H, Wang X, Ge X, Lu K, Jiang Y. Preventive hyperbaric oxygen therapy improves acute graft-versus-host disease by activating the Nrf2/HO-1 pathway. Front Immunol 2025; 16:1529176. [PMID: 40083556 PMCID: PMC11903425 DOI: 10.3389/fimmu.2025.1529176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Background Hyperbaric oxygen therapy (HBOT) has been confirmed as an effective and economical therapeutic modality for treating hemorrhagic cystitis (HC), whether induced by infection or acute graft-versus-host disease (aGVHD), in transplant recipients. However, its potential benefits in treating aGVHD remain largely unknown. This study explored the effects of HBOT on aGVHD and its underlying mechanisms. Methods The beneficial effects of HBOT on aGVHD were investigated in a murine model. Manifestations, pathological alterations, reactive oxygen species (ROS) levels in target organs, and survival data of the recipient mice were collected. Nuclear factor erythroid-derived 2-related factor 2 (Nrf2) and its downstream enzyme heme-oxygenase 1 (HO-1) expression in mouse samples were assessed via Western blot and immunohistochemistry analyses. ML385, an Nrf2 inhibitor, was used to validate the protective role of Nrf2 in the beneficial effect of HBOT on aGVHD. Furthermore, we initiated a clinical cohort study and collected data from the patients with definite aGVHD before and after HBOT to validate the preclinical conclusions. Results We found that HBOT alleviated aGVHD in mice, which was associated with a significantly prolonged overall survival (OS) and reduced pathological injury, whereas Nrf2 inhibition had the opposite effect. HBOT decreased ROS levels and proinflammatory cytokines, including IL-6 and TNF-α, while upregulated Nrf2 and its downstream antioxidant enzyme HO-1. In the clinical cohort study, the incidence of grades 1-3 aGVHD was significantly lower in the combination arm containing HBOT than in the HBOT-free cohort. Conclusion Preventive HBOT can mitigate aGVHD by activating the Nrf2/HO-1 signal transduction pathway, suggesting that HBOT may be a feasible approach for both the prevention and treatment of aGVHD. Clinical trial registration ClinicalTrials.gov, identifier NCT04502628.
Collapse
Affiliation(s)
- Chao Xue
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Hao Chen
- Department of Hyperbaric Oxygen Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yiou Zhao
- College of Life Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Dai Yuan
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mei Ding
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Huiting Qu
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- School of Medicine, Shandong University, Jinan, Shandong, China
| | - Xueling Ge
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Kang Lu
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yujie Jiang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
2
|
Choi JK, Mbanefo EC, Yadav MK, Alhakeem SA, Nagarajan V, Nunes NS, Kanakry CG, Egwuagu CE. Interleukin 35-producing B cells prolong the survival of GVHD mice by secreting exosomes with membrane-bound IL-35 and upregulating PD-1/LAG-3 checkpoint proteins. Theranostics 2025; 15:3610-3626. [PMID: 40093899 PMCID: PMC11905137 DOI: 10.7150/thno.105069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/18/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an effective treatment for aggressive hematologic malignancies. However, the risk of developing graft-versus-host disease (GVHD) is a significant barrier to allo-HSCT. GVHD is a debilitating condition with high mortality rates and current therapeutic options for GVHD are limited, with corticosteroids being the standard treatment. However, the adverse effects of steroids make prolonged use difficult, necessitating the development of safer therapies. IL-35-producing B-cells (i35-Bregs) have emerged as critical regulators of immunity during autoimmune diseases. In this study, we investigated whether i35-Bregs immunotherapy can suppress and mitigate GVHD. Methods: We administered a single dose of i35-Bregs (1.5×106) to mice undergoing allo-HSCT and monitored disease severity and survival of GVHD mice over 90 days post-transplantation. We discovered that i35-Bregs secrete exosomes containing membrane-bound IL-35 (i35-Exosomes) and investigated whether ex-vivo generated i35-exosomes can be used as stand-alone immunotherapy for GVHD. i35-Breg-induced expression of cytokines or checkpoint proteins (PD-1, LAG-3, CTLA-4) was analyzed by Flow cytometry, ELISA, and RNA-seq analysis. Characterization of membrane-bound IL-35 was by Proximity ligation assay (PLA), immunohistochemistry/Confocal microscopy and Alpha Fold-Multimer modeling. Results: A single dose of 1.5×106 i35-Breg reduced severity of GVHD and prolonged GVHD survival, with more than 70% i35-Breg-treated mice surviving beyond day-90 post-transplantation while observing 100% mortality among untreated mice by day-45. Contrary to the view that IL-35 is secreted cytokine, we show here that i35-Bregs mitigate GVHD via membrane-bound IL-35 and by secreting i35-exosomes. Furthermore, i35-Bregs or ex-vivo generated i35-exosomes induce alloreactive T-cells to upregulate checkpoint proteins associated with T-cell exhaustion and anergy, inhibiting alloreactive responses and propagating infectious-tolerance mechanisms that suppress GVHD. Importantly, i35-Bregs or i35-exosomes suppresses GVHD by increasing bystander lymphocytes coated with immunosuppressive i35-exosomes. Conclusions: This study demonstrates that i35-Bregs and i35-exosomes play a critical role in mitigating GVHD. The combination of i35-Breg and i35-exosome immunotherapy may be an effective strategy for treating GVHD and other inflammatory diseases.
Collapse
Affiliation(s)
- Jin Kyeong Choi
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States of America (USA)
- Department of Immunology, Jeonbuk National University Medical School, Jeonju, Jeonbuk, 54907, Republic of Korea
| | - Evaristus C. Mbanefo
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States of America (USA)
| | - Manoj Kumar Yadav
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States of America (USA)
| | - Sahar A. Alhakeem
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States of America (USA)
- Department of Biomedical Sciences, College of Health Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Vijayaraj Nagarajan
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States of America (USA)
| | - Natalia S. Nunes
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda MD 20892, United States of America
| | - Christopher G. Kanakry
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda MD 20892, United States of America
| | - Charles E. Egwuagu
- Molecular Immunology Section, Laboratory of Immunology, National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, United States of America (USA)
| |
Collapse
|
3
|
Li K, Feng J, Li M, Han L, Wu Y. Systematic Review of Interleukin-35 in Endothelial Dysfunction: A New Target for Therapeutic Intervention. Mediators Inflamm 2025; 2025:2003124. [PMID: 39974277 PMCID: PMC11839265 DOI: 10.1155/mi/2003124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial dysfunction is a significant factor in the pathogenesis of various diseases. In pathological states, endothelial cells (ECs) undergo activation, resulting in dysfunction characterized by the stimulation of inflammatory responses, oxidative stress, cell proliferation, blood coagulation, and vascular adhesions. Interleukin-35 (IL-35), a novel member of the IL-12 family, is primarily secreted by regulatory T cells (Tregs) and regulatory B cells (Bregs). The role of IL-35 in immunomodulation, antioxidative stress, resistance to apoptosis, control of EC activation, adhesion, and angiogenesis in ECs remains incompletely understood, as the specific mechanisms of IL-35 action and its regulation have yet to be fully elucidated. Therefore, this systematic review aims to comprehensively investigate the impact of IL-35 on ECs and their physiological roles in a range of conditions, including cardiovascular diseases, tumors, sepsis, and rheumatoid arthritis (RA), with the objective of elucidating the potential of IL-35 as a therapeutic target for these ailments.
Collapse
Affiliation(s)
- Kai Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Jie Feng
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| |
Collapse
|
4
|
Bakery HH, Hussein HAA, Ahmed OM, Abuelsaad ASA, Khalil RG. The potential therapeutic role of IL-35 in pathophysiological processes in type 1 diabetes mellitus. Cytokine 2024; 182:156732. [PMID: 39126765 DOI: 10.1016/j.cyto.2024.156732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/01/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
A chronic autoimmune condition known as type 1 diabetes mellitus (T1DM) has characteristics marked by a gradual immune-mediated deterioration of the β-cells that produce insulin and causes overt hyperglycemia. it affects more than 1.2 million kids and teenagers (0-19 years old). In both, the initiation and elimination phases of T1DM, cytokine-mediated immunity is crucial in controlling inflammation. T regulatory (Treg) cells, a crucial anti-inflammatory CD4+ T cell subset, secretes interleukin-35 (IL-35). The IL-35 has immunomodulatory properties by inhibiting pro-inflammatory cells and cytokines, increasing the secretion of interleukin-10 (IL-10) as well as transforming Growth Factor- β (TGF-β), along with stimulating the Treg and B regulatory (Breg) cells. IL-35, it is a possible target for cutting-edge therapies for cancers, inflammatory, infectious, and autoimmune diseases, including TIDM. Unanswered questions surround IL-35's function in T1DM. Increasing data suggests Treg cells play a crucial role in avoiding autoimmune T1DM. Throughout this review, we will explain the biological impacts of IL-35 and highlight the most recently progresses in the roles of IL-35 in treatment of T1DM; the knowledge gathered from these findings might lead to the development of new T1DM treatments. This review demonstrates the potential of IL-35 as an effective autoimmune diabetes inhibitor and points to its potential therapeutic value in T1DM clinical trials.
Collapse
Affiliation(s)
- Heba H Bakery
- Immunology Division, Faculty of Science, Beni-Suef University, Egypt
| | - Heba A A Hussein
- Faculty of Medicine, Egyptian Fellowship of Radiology, Beni-Suef University, Egypt
| | - Osama M Ahmed
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Egypt
| | | | - Rehab G Khalil
- Immunology Division, Faculty of Science, Beni-Suef University, Egypt.
| |
Collapse
|
5
|
Silva RCMC, Travassos LH, Dutra FF. The dichotomic role of single cytokines: Fine-tuning immune responses. Cytokine 2024; 173:156408. [PMID: 37925788 DOI: 10.1016/j.cyto.2023.156408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
Cytokines are known for their pleiotropic effects. They can be classified by their function as pro-inflammatory, such as tumor necrosis factor (TNF), interleukin (IL) 1 and IL-12, or anti-inflammatory, like IL-10, IL-35 and transforming growth factor β (TGF-β). Though this type of classification is an important simplification for the understanding of the general cytokine's role, it can be misleading. Here, we discuss recent studies that show a dichotomic role of the so-called pro and anti-inflammatory cytokines, highlighting that their function can be dependent on the microenvironment and their concentrations. Furthermore, we discuss how the back-and-forth interplay between cytokines and immunometabolism can influence the dichotomic role of inflammatory responses as an important target to complement cytokine-based therapies.
Collapse
Affiliation(s)
| | - Leonardo Holanda Travassos
- Laboratório de Receptores e Sinalização intracelular, Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil
| | - Fabianno Ferreira Dutra
- Laboratório de Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Huang A, Liu K, Yin Z, Liu J, Wei H, Xing S, Qu Y, Huang L, Li L, Li C, Zhang L, Li X, Zheng C, Liu Q, Jiang K. IL-35 Stabilizes Treg Phenotype to Protect Cardiac Allografts in Mice. Transplantation 2024; 108:161-174. [PMID: 37464473 PMCID: PMC10718222 DOI: 10.1097/tp.0000000000004707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Interleukin-35 (IL-35), secreted by regulatory T cells (Treg) and B cells, is immunosuppressive under both physiological and pathological conditions. However, the role of IL-35 in all responses has yet to be investigated. Here, we demonstrate that IL-35 protects allografts by stabilizing the Treg phenotype and suppressing CD8 + T-cell activation in a mouse heart transplantation model. METHODS The effect of IL-35 on immune cell infiltration in grafts and secondary lymphoid organs was examined using mass cytometry, flow cytometry, and immunofluorescence. Moreover, using quantitative real-time polymerase chain reaction, flow cytometry, and phospho-flow assays, we demonstrated that IL-35 maintains Treg phenotypes to restrain CD8 + T cells via the gp130/signal transducer and activator of transcription 1 pathway. RESULTS Mass cytometry analysis of intragraft immune cells showed that IL-35 decreased CD8 + T-cell infiltration and increased Foxp3 and IL-35 expressions in Treg. In vitro, we demonstrated that IL-35 directly promoted Treg phenotypic and functional stability and its IL-35 secretion, generating a positive feedback loop. However, Treg are required for IL-35 to exert its suppressive effect on CD8 + T cells in vitro. After depleting Treg in the recipient, IL-35 did not prolong graft survival or decrease CD8 + T-cell infiltration. Mechanistically, we found that IL-35 sustained Treg stability via the gp130/signal transducer and activator of transcription 1 signaling pathway. CONCLUSIONS Our findings highlight that IL-35 stabilizes the Treg phenotype to ameliorate CD8 + T-cell infiltration in the allograft, which has never been described in the transplanted immunological milieu.
Collapse
Affiliation(s)
- Ai Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kewei Liu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyi Yin
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Jie Liu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Hongyan Wei
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Shijie Xing
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Qu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liancheng Li
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Li
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Lei Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Xiaoshi Li
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Cunni Zheng
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Quan Liu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
A Rahman NA, Balasubramaniam VRMT, Yap WB. Potential of Interleukin (IL)-12 Group as Antivirals: Severe Viral Disease Prevention and Management. Int J Mol Sci 2023; 24:ijms24087350. [PMID: 37108513 PMCID: PMC10138811 DOI: 10.3390/ijms24087350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
The interleukin (IL)-12 family consists of pro- and anti-inflammatory cytokines that are able to signal the activation of host antiviral immunity while preventing over-reactive immune reactions due to active virus replication and viral clearance. Amongst others, IL-12 and IL-23 are produced and released by innate immune cells such as monocytes and macrophages to signal the proliferation of T cells and release of effector cytokines, which subsequently activate host defence against virus infections. Interestingly, the dualities of IL-27 and -35 are evidently shown in the course of virus infections; they regulate the synthesis of cytokines and antiviral molecules, proliferation of T cells, and viral antigen presentation in order to maximize virus clearance by the host immune system. In terms of anti-inflammatory reactions, IL-27 signals the formation of regulatory T cells (Treg) which in turn secrete IL-35 to control the scale of inflammatory response that takes place during virus infections. Given the multitasking of the IL-12 family in regards to the elimination of virus infections, its potential in antiviral therapy is unequivocally important. Thus, this work aims to delve deeper into the antiviral actions of the IL-12 family and their applications in antiviral therapies.
Collapse
Affiliation(s)
- Nur Azizah A Rahman
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Vinod R M T Balasubramaniam
- Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 46150, Malaysia
| | - Wei Boon Yap
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
- Biomedical Science Program, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
8
|
Lederer K, Maillard I. New mechanisms of GVHD suppression by Tregs. Blood 2023; 141:1655-1657. [PMID: 37022736 PMCID: PMC10113171 DOI: 10.1182/blood.2022019396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Affiliation(s)
| | - Ivan Maillard
- University of Pennsylvania Perelman School of Medicine
| |
Collapse
|
9
|
Zysk W, Gleń J, Trzeciak M. Current Insight into the Role of IL-35 and Its Potential Involvement in the Pathogenesis and Therapy of Atopic Dermatitis. Int J Mol Sci 2022; 23:ijms232415709. [PMID: 36555351 PMCID: PMC9779445 DOI: 10.3390/ijms232415709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Interleukin 35 (IL-35), a new member of the IL-12 family of heterodimeric cytokines, could induce two different types of regulatory cells including regulatory T and B cells such as IL-35-induced regulatory T cells and IL-10-producing regulatory B cells (IL-10+Bregs), and IL-35-producing regulatory B cells (IL-35+Bregs). These cells appear to play an important role in modulating the immune system in numerous diseases. Several findings suggested that the expression of IL-35 is dysregulated in many autoimmune, inflammatory, and allergic diseases. Due to the functions of IL-35, it seems that this cytokine may act as an efficient therapeutic strategy for numerous conditions including atopic dermatitis (AD). We aimed to provide a comprehensive overview of the role of IL-35 in modulating the immune system. Additionally, we highlight IL-35 as a specific immunological target, discuss its possible involvement in the pathogenesis of AD, and hypothesize that IL-35 may become a novel target for the treatment of AD. However, further studies are required to evaluate this hypothesis.
Collapse
Affiliation(s)
- Weronika Zysk
- Dermatological Students Scientific Association, Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland
| | - Jolanta Gleń
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland
| | - Magdalena Trzeciak
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland
- Correspondence: ; Tel.: +48-58-584-40-10
| |
Collapse
|
10
|
Dendritic cell-derived IL-27 p28 regulates T cell program in pathogenicity and alleviates acute graft-versus-host disease. Signal Transduct Target Ther 2022; 7:319. [PMID: 36109504 PMCID: PMC9477797 DOI: 10.1038/s41392-022-01147-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 06/30/2022] [Accepted: 07/29/2022] [Indexed: 11/18/2022] Open
Abstract
Interleukin 27 (IL-27), a heterodimeric cytokine composed of Epstein-Barr virus-induced 3 and p28, is a pleiotropic cytokine with both pro-and anti-inflammatory properties. However, the precise role of IL-27 in acute graft-versus-host disease is not yet fully understood. In this study, utilizing mice with IL-27 p28 deficiency in dendritic cells (DCs), we demonstrated that IL-27 p28 deficiency resulted in impaired Treg cell function and enhanced effector T cell responses, corresponding to aggravated aGVHD in mice. In addition, using single-cell RNA sequencing, we found that loss of IL-27 p28 impaired Treg cell generation and promoted IL-1R2+TIGIT+ pathogenic CD4+ T cells in the thymus at a steady state. Mechanistically, IL-27 p28 deficiency promoted STAT1 phosphorylation and Th1 cell responses, leading to the inhibition of Treg cell differentiation and function. Finally, patients with high levels of IL-27 p28 in serum showed a substantially decreased occurrence of grade II-IV aGVHD and more favorable overall survival than those with low levels of IL-27 p28. Thus, our results suggest a protective role of DC-derived IL-27 p28 in the pathogenesis of aGVHD through modulation of the Treg/Teff cell balance during thymic development. IL-27 p28 may be a valuable marker for predicting aGVHD development after transplantation in humans.
Collapse
|
11
|
Elevated Plasma Interleukin-35 as a Prognostic Indicator in Localized Clear Cell Renal Cell Carcinoma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6886590. [PMID: 36124013 PMCID: PMC9482474 DOI: 10.1155/2022/6886590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/10/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022]
Abstract
Purpose The aim of the study is to investigate the prognostic value of plasma interleukin-35 in the surgical treatment of patients with clear cell renal cell carcinoma (ccRCC). Material and Methods. Plasma IL-35 levels were measured in patients with ccRCC. The cut-off value of IL-35 was determined by the receiver operating characteristic (ROC) analysis and the area under the curve (AUC). The effects of the IL-35 and other clinicopathological characteristics on overall survival (OS) and progression-free survival (PFS) were evaluated using the univariate and multivariate logistic regression analysis. Result Sixty-four ccRCC patients admitted to the urology department at the First Affiliated Hospital of Soochow University were selected, of whom 50 were diagnosed with localized ccRCC. Plasma interleukin-35 levels were significantly higher in patients with ccRCC than that in healthy controls. The cut-off value of IL-35 was 99.7 pg/mL. Multivariate analysis selected by univariate analyses demonstrated that the preoperative IL-35 was an independent prognostic factor for 5-year OS (OR: 1.02, 95% CI: 1.01 to 1.04, p < 0.0001) and 5-year PFS (OR: 1.02, 95% CI: 1.00 to 1.03, p=0.011) in all patients with localized ccRCC. Conclusion Current results indicate that preoperative IL-35 is an independent prognostic marker for OS and RFS in patients with localized ccRCC after surgery.
Collapse
|
12
|
Tong X, Ru Y, Fu J, Wang Y, Zhu J, Ding Y, Lv F, Yang M, Wei X, Liu C, Liu X, Lei L, Wu X, Guo L, Xu Y, Li J, Wu P, Gong H, Chen J, Wu D. Fucosylation Promotes Cytolytic Function and Accumulation of NK Cells in B Cell Lymphoma. Front Immunol 2022; 13:904693. [PMID: 35784355 PMCID: PMC9240281 DOI: 10.3389/fimmu.2022.904693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/11/2022] [Indexed: 12/17/2022] Open
Abstract
Natural killer (NK) cells have been demonstrated as a promising cellular therapy as they exert potent anti-tumor immune responses. However, applications of NK cells to tumor immunotherapy, especially in the treatment of advanced hematopoietic and solid malignancies, are still limited due to the compromised survival and short persistence of the transferred NK cells in vivo. Here, we observed that fucosyltransferase (FUT) 7 and 8 were highly expressed on NK cells, and the expression of CLA was positively correlated with the accumulation of NK cells in clinical B cell lymphoma development. Via enzyme-mediated ex vivo cell-surface fucosylation, the cytolytic effect of NK cells against B cell lymphoma was significantly augmented. Fucosylation also promoted NK cell accumulation in B cell lymphoma-targeted tissues by enhancing their binding to E-selectin. Moreover, fucosylation of NK cells also facilitated stronger T cell anti-tumor immune responses. These findings suggest that ex vivo fucosylation contributes to enhancing the effector functions of NK cells and may serve as a novel strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Xing Tong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuhua Ru
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jianhong Fu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Ying Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jinjin Zhu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Yiyang Ding
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Fulian Lv
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Menglu Yang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xiya Wei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Chenchen Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xin Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Lei Lei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xiaojin Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jie Li
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
- *Correspondence: Peng Wu, ; Huanle Gong, ; Jia Chen, ; Depei Wu,
| | - Huanle Gong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Peng Wu, ; Huanle Gong, ; Jia Chen, ; Depei Wu,
| | - Jia Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Peng Wu, ; Huanle Gong, ; Jia Chen, ; Depei Wu,
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Peng Wu, ; Huanle Gong, ; Jia Chen, ; Depei Wu,
| |
Collapse
|
13
|
Lv K, Hu B, Xu M, Wan L, Jin Z, Xu M, Du Y, Ma K, Lv Q, Xu Y, Lei L, Gong H, Liu H, Wu D, Liu Y. IL-39 promotes chronic graft-versus-host disease by increasing T and B Cell pathogenicity. Exp Hematol Oncol 2022; 11:34. [PMID: 35655245 PMCID: PMC9161463 DOI: 10.1186/s40164-022-00286-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic graft-versus-host disease (cGVHD) remains a major complication during the late phase of allogeneic hematopoietic stem cell transplantation (allo-HSCT). IL-39, a newly described pro-inflammatory cytokine belonging to the IL-12 family, plays a role in lupus development. Recently, IL-39 has been identified as a pathogenic factor in acute GVHD (aGVHD). However, the role of IL-39 in the pathogenesis of cGVHD remains unclear. METHODS We constructed a recombinant IL-39 plasmid and established scleroderma and lupus-like cGVHD models. Quantitative PCR and enzyme-linked immunosorbent assay (ELISA) were used to detect IL-39 expression in mice and patients post transplantation, respectively. Hydrodynamic gene transfer (HGT) was performed to achieve IL-39 overexpression in vivo. Multiparameter flow cytometry, western blotting, and assays in vitro were performed to investigate the effect of IL-39 on cGVHD. RESULTS The relative expression of IL-23p19 and EBi3 was significantly increased in the intestine of cGVHD mice on day 40 post allo-HSCT, and IL-39 levels were significantly elevated in the serum of patients following allo-HSCT. Overexpression of IL-39 significantly aggravated the severity of cGVHD. Increased IL-39 levels promoted T-cell activation and germinal center responses, and may exacerbate thymic damage. Consistently, blocking IL-39 markedly ameliorated immune dysregulation in the cGVHD mice. Furthermore, we found that IL-39 was produced by B cells, CD11b+ cells, and CD8+T cells after activation. Stimulation of IL-39 led to upregulation of the IL-39 receptor on CD4+T cells and further caused activation of the STAT1/STAT3 pathway, through which IL-39 may exert its pro-inflammatory effects. CONCLUSION Our study reveals a critical role for IL-39 in cGVHD pathogenesis and indicates that IL-39 may serve as a potential therapeutic target for cGVHD prevention.
Collapse
Affiliation(s)
- Kangkang Lv
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Bo Hu
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Mingzhu Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Li Wan
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziqi Jin
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Mimi Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuanyuan Du
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Kunpeng Ma
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Quansheng Lv
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Lei
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Huanle Gong
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haiyan Liu
- Department of Microbiology and Immunology, Life Sciences Institute, Immunology Translational Research ProgramYong Loo Lin School of MedicineImmunology ProgramNational University of Singapore, Singapore, Singapore.
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, China. .,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Yuejun Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, China. .,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
14
|
Lombardelli L, Logiodice F, Kullolli O, Haller H, Agostinis C, Bulla R, Rukavina D, Piccinni MP. At Embryo Implantation Site IL-35 Secreted by Trophoblast, Polarizing T Cells towards IL-35+ IL-10+ IL-4+ Th2-Type Cells, Could Favour Fetal Allograft Tolerance and Pregnancy Success. Int J Mol Sci 2022; 23:ijms23094926. [PMID: 35563316 PMCID: PMC9103079 DOI: 10.3390/ijms23094926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/03/2022] Open
Abstract
We investigated the role of rhIL-35, at low concentrations compatible with those produced by human trophoblast cells (less than 1 ng/mL), on human T helper (Th) cell functions and the presence of decidual IL-35-producing Th cells in human pregnancy. We found that human trophoblast cells produced IL-35 but not IL-4 or IL-10. RhIL-35, at concentrations produced by human trophoblasts, polarized T cells towards IL-35+, IL-10+, IL-4+ Th2-type cells and to Foxp3+ EBI3+ p35+ T reg cells producing IL-35 but not IL-10 and IL-4. Moreover, rhIL-35 at low concentrations did not suppress the proliferation of Th cells but stimulated IL-4 and IL-10 production by established Th clones. In particular, Th1-type clones acquired the capacity to produce IL-4. In addition, purified human trophoblast cell supernatants containing IL-35 upregulated IL-4 and IL-10 production by Th clones. Finally, IL-35+, IL-10+, IL-4+ Th2-type cells, which were found to be induced by low concentrations of IL-35 compatible with those produced by human trophoblasts, are exclusively present in the decidua of a successful pregnancy and at the embryo implantation site, suggesting their stringent dependence on trophoblast cells. Thus, the proximity of Th cells to IL-35-producing trophoblasts could be the determining factor for the differentiation of IL-35+, IL-10+, IL-4+ Th2-type cells that are crucial for human pregnancy success.
Collapse
Affiliation(s)
- Letizia Lombardelli
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.L.); (F.L.); (O.K.)
| | - Federica Logiodice
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.L.); (F.L.); (O.K.)
| | - Ornela Kullolli
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.L.); (F.L.); (O.K.)
| | - Herman Haller
- Department of Gynecology and Obstetrics, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia;
| | - Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Daniel Rukavina
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia;
| | - Marie-Pierre Piccinni
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.L.); (F.L.); (O.K.)
- Correspondence: ; Tel.: +39-055-275-8338
| |
Collapse
|
15
|
Torabijahromi M, Roozbeh J, Raeesjalali G, Shafiee M, Rasaei N, Heidari M, Karimi MH. Comparison of FOXP3 and Interleukin 35 Expression Profiles in Kidney Transplant Recipients With Excellent Long-Term Graft Function and Acute Rejection. EXP CLIN TRANSPLANT 2021; 19:1142-1148. [PMID: 34812705 DOI: 10.6002/ect.2021.0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Transplant tolerance is defined as graft acceptance without long-term use of immunosuppressive agents. Regulatory T cells are involved in the maintenance of peripheral self-tolerance by actively suppressing the activation and expansion of autoreactive T cells. In the present study, we compared the expression profiles of forkhead box protein P3 (FOXP3) and interleukin 35 in kidney transplant recipients who had excellent long-term graft function under immunosuppression versus recipients who had acute rejection. MATERIALS AND METHODS The 40 kidney transplant recipients included in this study were divided into 2 groups: 27 recipients with excellent long-term graft function and 13 recipients with acute rejection. After collection of whole peripheral blood, peripheral blood mononuclear cells were isolated from the blood samples. After RNAextraction and cDNAsynthesis from each collected sample, expression levels of interleukin 35 and FOXP3 were determined using in-house SYBER green-based real-time polymerase chain reaction. We used t tests to analyze data. RESULTS Mean ages of recipients with excellent longterm graft function and recipients with acute rejection were 42.1 and 45.5 years, respectively. We found that FOXP3 and interleukin 35 expression levels were significantly increased in recipients with excellentlongterm graftfunction comparedwith recipientswith acute rejection. FOXP3 expression levels were significantly higher in those with excellent long-term graft function with graft survivalrate of <10 years,whereas interleukin 35 expression levels were significantly higher in patients with graft survival rate >10 years (P < .05). Expression levels of FOXP3 and interleukin 35 were greater in those from 35 to 50 years old versus with those in the other age ranges. CONCLUSIONS Expression patterns of FOXP3 and interleukin 35 may have the potential to be used as prognostic biomarkers for kidney transplant outcomes.
Collapse
Affiliation(s)
- Mahsa Torabijahromi
- From the Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | | | | | |
Collapse
|
16
|
Hill GR, Betts BC, Tkachev V, Kean LS, Blazar BR. Current Concepts and Advances in Graft-Versus-Host Disease Immunology. Annu Rev Immunol 2021; 39:19-49. [PMID: 33428454 PMCID: PMC8085043 DOI: 10.1146/annurev-immunol-102119-073227] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Worldwide, each year over 30,000 patients undergo an allogeneic hema-topoietic stem cell transplantation with the intent to cure high-risk hematologic malignancy, immunodeficiency, metabolic disease, or a life-threatening bone marrow failure syndrome. Despite substantial advances in donor selection and conditioning regimens and greater availability of allograft sources, transplant recipients still endure the morbidity and mortality of graft-versus-host disease (GVHD). Herein, we identify key aspects of acute and chronic GVHD pathophysiology, including host/donor cell effectors, gut dysbiosis, immune system and cytokine imbalance, and the interface between inflammation and tissue fibrosis. In particular, we also summarize the translational application of this heightened understanding of immune dysregulation in the design of novel therapies to prevent and treat GVHD.
Collapse
Affiliation(s)
- Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA;
- Division of Medical Oncology University of Washington, Seattle, Washington 98109, USA
| | - Brian C Betts
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Victor Tkachev
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; ,
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; ,
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, USA;
| |
Collapse
|
17
|
Song Y, Zhu Y, Hu B, Liu Y, Lin D, Jin Z, Yin Z, Dong C, Wu D, Liu H. Donor γδT Cells Promote GVL Effect and Mitigate aGVHD in Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2020; 11:558143. [PMID: 33178187 PMCID: PMC7596318 DOI: 10.3389/fimmu.2020.558143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/22/2020] [Indexed: 01/03/2023] Open
Abstract
Disease relapse and graft-versus-host disease (GVHD) are the major complications affecting the outcomes of allogeneic hematopoietic stem cell transplantation (allo-HSCT). While the functions of αβT cells are extensively studied, the role of donor γδT cells in allo-HSCT is less well defined. Using TCRδ-/- donors lacking γδT cells, we demonstrated that donor γδT cells were critical in mediating graft-versus-leukemia (GVL) effect during allo-HSCT. In the absence of donor γδT cells, IFN-γ production by CD8+ T cells was severely impaired. Vγ4 subset was the major γδT cell subset mediating the GVL effect in vivo, which was partially dependent on IL-17A. Meanwhile, donor γδT cells could mitigate acute GVHD in a murine allo-HSCT model by suppressing CD4+ T cell activation and the major γδT cell subset that exerted this protective function was also Vγ4 γδT cells. Therefore, our findings provide evidence that donor γδT cells, especially Vγ4 subset, can enhance GVL effect and mitigate aGVHD during allo-HSCT.
Collapse
Affiliation(s)
- Yuan Song
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ying Zhu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Bo Hu
- Institute of Blood and Marrow Transplantation, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, Soochow University, Suzhou, China
| | - Yonghao Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dandan Lin
- Institute of Blood and Marrow Transplantation, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, Soochow University, Suzhou, China
| | - Ziqi Jin
- Institute of Blood and Marrow Transplantation, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, Soochow University, Suzhou, China
| | - Zhinan Yin
- Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Jinan University, Zhuhai, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Depei Wu
- Institute of Blood and Marrow Transplantation, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Diseases, Soochow University, Suzhou, China
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
18
|
Zhang P, Hill GR. Interleukin-10 mediated immune regulation after stem cell transplantation: Mechanisms and implications for therapeutic intervention. Semin Immunol 2019; 44:101322. [PMID: 31640914 DOI: 10.1016/j.smim.2019.101322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/08/2019] [Indexed: 12/23/2022]
Abstract
Interleukin-10 (IL-10) is a multi-faceted anti-inflammatory cytokine which plays an essential role in immune tolerance. Indeed, deficiency of IL-10 or its receptor results in aberrant immune responses that lead to immunopathology. Graft-versus-host disease (GVHD) is the limiting complication of allogeneic stem cell transplantation (SCT) and results from an imbalance in pathological versus regulatory immune networks. A number of immune cells exert their immunomodulatory role through secretion of IL-10 or induction of IL-10-secreting cells after SCT. Type-1 regulatory T cells (Tr1 cells) and FoxP3+ regulatory T cells (Tregs) are predominant sources of IL-10 after SCT and the critical role of this cytokine in preventing GVHD is now established. Recently, intriguing interactions among IL-10, immune cells, commensal microbes and host tissues in the gastrointestinal (GI) tract and other barrier surfaces have been uncovered. We now understand that IL-10 secretion is dynamically modulated by the availability of antigen, co-stimulatory signals, cytokines, commensal microbes and their metabolites in the microenvironment. In this review, we provide an overview of the control of IL-10 secretion and signaling after SCT and the therapeutic interventions, with a focus on Tr1 cells.
Collapse
Affiliation(s)
- Ping Zhang
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Division of Medical Oncology, The University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
19
|
Xue W, Yan D, Kan Q. Interleukin-35 as an Emerging Player in Tumor Microenvironment. J Cancer 2019; 10:2074-2082. [PMID: 31205568 PMCID: PMC6548173 DOI: 10.7150/jca.29170] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 03/05/2019] [Indexed: 12/15/2022] Open
Abstract
IL-35 is the newest member of IL-12 family. A dimeric protein consisting of two separate subunits has manifested suppressive actions on immune system, which is counterproductive in the context of cancers. Various reports have confirmed its inhibitory role on immune system which is carried out via formation of IL-35-producing regulatory T cells (iTr35), increased Treg development and suppressive Th17 cells growth. Although last decade has seen a great deal of scientific interest on this subject, the exact role, precise signal transduction and elaborative functions of IL-35 in tumor microenvironment (TME) remained elusive. Search for anti-IL-35 therapies have exhibited limited success in animal models. Contrarily, few studies have denied the idea that IL-35 plays a role in cancer. The purpose of this review is to analyze the reported scientific data on continuous symphony of IL-35 in cancers since the inception of former.
Collapse
Affiliation(s)
- Wenhua Xue
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Dan Yan
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Quancheng Kan
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Bastian D, Wu Y, Betts BC, Yu XZ. The IL-12 Cytokine and Receptor Family in Graft-vs.-Host Disease. Front Immunol 2019; 10:988. [PMID: 31139181 PMCID: PMC6518430 DOI: 10.3389/fimmu.2019.00988] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/16/2019] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is performed with curative intent for high- risk blood cancers and bone marrow failure syndromes; yet the development of acute and chronic graft-vs.-host disease (GVHD) remain preeminent causes of death and morbidity. The IL-12 family of cytokines is comprised of IL-12, IL-23, IL-27, IL-35, and IL-39. This family of cytokines is biologically distinct in that they are composed of functional heterodimers, which bind to cognate heterodimeric receptor chains expressed on T cells. Of these, IL-12 and IL-23 share a common β cytokine subunit, p40, as well as a receptor chain: IL-12Rβ1. IL-12 and IL-23 have been documented as proinflammatory mediators of GVHD, responsible for T helper 1 (Th1) differentiation and T helper 17 (Th17) stabilization, respectively. The role of IL-27 is less defined, seemingly immune suppressive via IL-10 secretion by Type 1 regulatory (Tr1) cells yet promoting inflammation through impairing CD4+ T regulatory (Treg) development and/or enhancing Th1 differentiation. More recently, IL-35 was described as a potent anti-inflammatory agent produced by regulatory B and T cells. The role of the newest member, IL-39, has been implicated in proinflammatory B cell responses but has not been explored in the context of allo-HCT. This review is directed at discussing the current literature relevant to each IL-12-family cytokine and cognate receptor engagement, as well as the consequential downstream signaling implications, during GVHD pathogenesis. Additionally, we will provide an overview of translational strategies targeting the IL-12 family cytokines, their receptors, and subsequent signal transduction to control GVHD.
Collapse
Affiliation(s)
- David Bastian
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Brian C Betts
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
21
|
Tang L, Ma S, Gong H, Wang J, Xu Y, Wu D, Sun A. PD-L1 Ameliorates Murine Acute Graft-Versus-Host Disease by Suppressing Effector But Not Regulatory T Cells Function. Arch Immunol Ther Exp (Warsz) 2019; 67:179-187. [PMID: 30927016 DOI: 10.1007/s00005-019-00539-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 03/18/2019] [Indexed: 01/09/2023]
Abstract
There is increasing evidence that interaction between programmed death 1 (PD-1) and its ligands PD-1 (PD-L1) plays a critical role in the pathology of acute graft-versus-host disease (aGVHD). However, the role of PD-L1 in the development of aGVHD has been controversial in recent mouse studies. In this study, we carried out studies in a murine aGVHD model to clarify the role of PD-L1 in aGVHD pathogenesis. We found that systemic overexpression of PD-L1 by hydrodynamic gene transfer (HGT) method in vivo ameliorates aGVHD-induced lethality in mice. Systemic overexpression of PD-L1 inhibits the donor T cells activation, effector memory status, as well as Th1 and Th17 cells responses in vivo. In addition, PD-L1 Ig treatment significantly suppressed T cells' proliferation, promoted T cells' apoptosis, and reduced pro-inflammatory cytokines expression by effector T cells in vitro in the stimulation of anti-CD3/CD28 and allogeneic dendritic cells. However, we found that PD-L1 overexpression did not affect Treg cells' differentiation in vivo and in vitro, depletion of Treg cells in PD-L1 HGT recipients did not aggravate aGVHD mortality. Therefore, our results demonstrated that systemic treatment with PD-L1 protein ameliorates aGVHD by suppressing effector but not regulatory T cell function. Our findings suggest that systemic treatment with PD-L1 may be a potential strategy to prevent or ameliorate aGVHD.
Collapse
Affiliation(s)
- Lin Tang
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China.,Department of Hematology, Changzhou Traditional Chinese Medicine Hospital, Changzhou, 213003, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
| | - Shoubao Ma
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China. .,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China.
| | - Huanle Gong
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
| | - Jun Wang
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
| | - Yang Xu
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China.,Department of Hematology, Changzhou Traditional Chinese Medicine Hospital, Changzhou, 213003, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China
| | - Depei Wu
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China. .,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China.
| | - Aining Sun
- Department of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China. .,Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, 215123, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006, China.
| |
Collapse
|
22
|
Wang W, Zhao N, Li B, Gao H, Yan Y, Guo H. Inhibition of cardiac allograft rejection in mice using interleukin-35-modified mesenchymal stem cells. Scand J Immunol 2019; 89:e12750. [PMID: 30664805 DOI: 10.1111/sji.12750] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/14/2019] [Accepted: 01/16/2019] [Indexed: 02/03/2023]
Abstract
Interleukin-35 (IL-35) is a cytokine recently discovered to play a potent immunosuppressive role by intensifying the functions of regulatory T cells and inhibiting the proliferation and functions of T helper 1 and T helper 17 cells. Mesenchymal stem cells (MSCs) have recently emerged as promising candidates for cell-based immune therapy, and our previous study showed that IL-35 gene modification can effectively enhance the therapeutic effect of MSCs in vitro. In this study, we isolated adipose tissue-derived MSCs in vitro and infected them with lentiviral vectors overexpressing the IL-35 gene, thereby creating IL-35-MSCs. Subsequently, IL-35-MSCs were then injected into mice of the allogeneic heterotopic abdominal heart transplant model to determine their effect on allograft rejection. The results showed that IL-35-MSCs could continuously secrete IL-35 in vivo and in vitro, successfully alleviate allograft rejection and prolong graft survival. In addition, compared to MSCs, IL-35-MSCs showed a stronger immunosuppressive ability and further reduced the percentage of Th17 cells, increased the proportion of CD4+ Foxp3+ T cells, and regulated Th1/Th2 balance in heart transplant mice. These findings suggest that IL-35-MSCs have more advantages than MSCs in inhibiting graft rejection and may thus provide a new approach for inducing immune tolerance during transplantation.
Collapse
Affiliation(s)
- Wei Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Na Zhao
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Baozhu Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Haopeng Gao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yongjia Yan
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Guo
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
23
|
Chang YJ, Zhao XY, Huang XJ. Strategies for Enhancing and Preserving Anti-leukemia Effects Without Aggravating Graft-Versus-Host Disease. Front Immunol 2018; 9:3041. [PMID: 30619371 PMCID: PMC6308132 DOI: 10.3389/fimmu.2018.03041] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/10/2018] [Indexed: 12/29/2022] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) is a curable method for the treatment of hematological malignancies. In the past two decades, the establishment of haploidentical transplant modalities make “everyone has a donor” become a reality. However, graft-versus-host disease (GVHD) and relapse remain the major two causes of death either in the human leukocyte antigen (HLA)-matched transplant or haploidentical transplant settings, both of which restrict the improvement of transplant outcomes. Preclinical mice model showed that both donor-derived T cells and natural killer (NK) cells play important role in the pathogenesis of GVHD and the effects of graft-versus-leukemia (GVL). Hence, understanding the immune mechanisms of GVHD and GVL would provide potential strategies for the control of leukemia relapse without aggravating GVHD. The purpose of the current review is to summarize the biology of GVHD and GVL responses in preclinical models and to discuss potential novel therapeutic strategies to reduce the relapse rate after allo-SCT. We will also review the approaches, including optimal donor selection and, conditioning regimens, donor lymphocyte infusion, BCR/ABL-specific CTL, and chimeric antigen receptor-modified T cells, which have been successfully used in the clinic to enhance and preserve anti-leukemia activity, especially GVL effects, without aggravating GVHD or alleviate GVHD.
Collapse
Affiliation(s)
- Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
24
|
Gong H, Ma S, Liu S, Liu Y, Jin Z, Zhu Y, Song Y, Lei L, Hu B, Mei Y, Liu H, Liu Y, Wu Y, Dong C, Xu Y, Wu D, Liu H. IL-17C Mitigates Murine Acute Graft-vs.-Host Disease by Promoting Intestinal Barrier Functions and Treg Differentiation. Front Immunol 2018; 9:2724. [PMID: 30534126 PMCID: PMC6275224 DOI: 10.3389/fimmu.2018.02724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 11/05/2018] [Indexed: 12/19/2022] Open
Abstract
Acute graft-vs.-host disease (aGVHD) is one of the major complications and results in high mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). IL-17C is involved in many inflammatory immune disorders. However, the role of IL-17C in aGVHD remains unknown. Here we demonstrated that IL-17C deficiency in the graft significantly promoted alloreactive T cell responses and induced aggravated aGVHD compared with wildtype donors in a fully MHC-mismatched allo-HSCT model. In contrast, IL-17C overexpression ameliorated aGVHD. IL-17C deficiency increased intestinal epithelial permeability and elevated inflammatory cytokine production, leading to an enhanced aGVHD progression. Tregs was reduced in recipients of IL-17C−/− graft, whilst restored after IL-17C overexpression. Decreased Treg differentiation was abrogated after neutralizing IFN-γ, but not IL-6. Moreover, depletion of Tregs diminished the protective effect of IL-17C. Of note, patients with low IL-17C expression displayed higher aGVHD incidence together with poor overall survival, thereby IL-17C could be an independent risk factor for aGVHD development. Our results are the first demonstrating the protective role of IL-17C in aGVHD by promoting intestinal barrier functions and Treg differentiation in a MHC fully mismatched murine aGVHD model. IL-17C may serve as a novel biomarker and potential therapeutic target for aGVHD.
Collapse
Affiliation(s)
- Huanle Gong
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shoubao Ma
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shuangzhu Liu
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yonghao Liu
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziqi Jin
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Zhu
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuan Song
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Lei Lei
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bo Hu
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Mei
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Hong Liu
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuejun Liu
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Wu
- School of Radiation Medicine and Protection School for Radiological and Interdisciplinary Science, Soochow University, Suzhou, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Yang Xu
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Guo H, Li B, Wang W, Zhao N, Gao H. Mesenchymal stem cells overexpressing IL-35: a novel immunosuppressive strategy and therapeutic target for inducing transplant tolerance. Stem Cell Res Ther 2018; 9:254. [PMID: 30257721 PMCID: PMC6158805 DOI: 10.1186/s13287-018-0988-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Inducing donor-specific immunological tolerance, which avoids the complications of long-term immunosuppression, is an important goal in organ transplantation. Interleukin-35 (IL-35), a cytokine identified in 2007, is mainly secreted by regulatory T cells (Tregs) and is essential for Tregs to exert their maximal immunoregulatory activity in vitro and in vivo. A growing number of studies show that IL-35 plays an important role in autoimmune diseases and infectious diseases. Recent research has shown that IL-35 could effectively alleviate allograft rejection and has the potential to be a novel therapeutic strategy for graft rejection. With increasing study of immunoregulation, cell-based therapy has become a novel approach to attenuate rejection after transplantation. Mesenchymal stem cells (MSCs), which exhibit important properties of multilineage differentiation, tissue repair, and immunoregulation, have recently emerged as attractive candidates for cell-based therapeutics, especially in transplantation. Accumulating evidence demonstrates that the therapeutic abilities of MSCs can be amplified by gene modification. Therefore, researchers have constructed IL-35 gene-modified MSCs and explored their functions and mechanisms in some disease models. In this review, we discuss the potential tolerance-inducing effects of MSCs in transplantation and briefly introduce the immunoregulatory functions of the IL-35 gene-modified MSCs.
Collapse
Affiliation(s)
- Hao Guo
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Baozhu Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Wei Wang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Na Zhao
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Haopeng Gao
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| |
Collapse
|
26
|
Zhang X, Zhang Z, He Z, Ju M, Li J, Yuan J, Jing Y, Li K, Liu Y, Li G. Interleukin 35 induced Th2 and Tregs bias under normal conditions in mice. PeerJ 2018; 6:e5638. [PMID: 30258726 PMCID: PMC6152461 DOI: 10.7717/peerj.5638] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022] Open
Abstract
Objective The benefits of IL-35 treatment have been verified in multiple animal models of diseases, while its influence on T cells immunity under normal condition still needs to be elucidated. The present study was designed to investigate the effects modulating IL-35 levels in vivo and in vitro on T cells, response and also the effects on T cells subsets in normal mice. Methods A plasmid pMSCV-IL-35-GFP carrying mouse linear IL-35 fragment with two subunits joint together was constructed and the heterodimer expression was confirmed. Normal mice were randomly divided into three groups and received an intravenous injection of PBS, pMSCV-GFP and pMSCV-IL-35-GFP respectively. After 72 h, spleen tissues and peripheral blood were harvested for following analysis. Meanwhile, splenic T cells were isolated and incubated with 10, 30, or 50 ng/mL recombinant IL-35 factor for 24 h with the addition of anti-CD3/CD28 in vitro. T-cell subsets were assessed by Fluorescence activated cell sorting (FACS) and related cytokines together with effector molecules were determined by real time PCR. Results Western blotting confirmed a 52 kDa band in the cell lysate of HEK 293T transducted with pMSCV-IL-35-GFP plasmid, indicating a successful expression of IL-35. Ebi3 and IL-12A, two subunits of IL-35, could be identified 72 h post DNA injection. IL-35 upregulation in vivo effectively inhibit CD4+ and CD8+ T cell proliferation and Th1 cytokine secretion. Effector molecules of CD8+ T cells were also remarkably suppressed. On the contrary, high level of IL-35 significantly induced CD4+ CD25+ Tregs and Th2 enhancement. The in vitro study provided similar results. Conclusion The results indicated Th1 and CD8+ T cell inhibition and Th2 and Tregs bias in the presence of IL-35 under a normal state which partly contributed to its therapeutic potential.
Collapse
Affiliation(s)
- Xiaoning Zhang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhiqiang Zhang
- Department of Pathology, Tianjin Hospital of ITCWM, Nankai Hospital, Tianjin, China
| | - Zhiqiang He
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mingyan Ju
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jiaci Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jinghua Yuan
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yaqing Jing
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Keqiu Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yi Liu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Guang Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
27
|
Hu L, Chen C, Zhang J, Wu K, Zhang X, Liu H, Hou J. IL-35 Pretreatment Alleviates Lipopolysaccharide-Induced Acute Kidney Injury in Mice by Inhibiting NF-κB Activation. Inflammation 2018; 40:1393-1400. [PMID: 28497278 DOI: 10.1007/s10753-017-0582-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Septic acute kidney injury (AKI) is a public health problem with high mortality. Suppression of over-active inflammation is considered as a promising strategy for septic AKI. In this study, we evaluated the prophylactic effect of interleukin (IL)-35, the unique immune-suppressive member of IL-12 cytokine family, on lipopolysaccharide (LPS)-induced AKI in mice, and found that compared with control mice given empty vector, mice pretreated with plasmid encoding IL-35 (pIL-35) significantly improved renal function indicated by reduced blood urea nitrogen (BUN) and serum creatinine (SCr), and obviously alleviated renal pathological changes. To explore the underlying protective mechanisms, we found that pIL-35 treatment could robustly reduce the production of renal pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), with no significant impact on IL-10, an anti-inflammatory cytokine. Furthermore, our results revealed that IL-35 pretreatment could potentially inhibit the activation of renal NF-κB signaling pathway in LPS-induced AKI mice. Taken together, our study indicated that IL-35 pretreatment could efficiently prevent LPS-induced AKI via inhibiting NF-κB activation and reducing pro-inflammatory cytokine production, and it might represent a novel therapeutic strategy against septic AKI and other inflammatory renal diseases.
Collapse
Affiliation(s)
- Linkun Hu
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Rd, Suzhou, 215006, People's Republic of China
| | - Cheng Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Rd, Suzhou, 215006, People's Republic of China
| | - Jun Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Rd, Suzhou, 215006, People's Republic of China
| | - Kerong Wu
- Center of Uro-nephrological Disease, Ningbo First Hospital, NO. 59 Liuting Avenue, Haishu District, Ningbo, 315000, People's Republic of China
| | - Xuefeng Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Rd, Suzhou, 215006, People's Republic of China
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117456, Singapore.
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Rd, Suzhou, 215006, People's Republic of China.
| |
Collapse
|
28
|
Song Y, Hu B, Liu Y, Jin Z, Zhang Y, Lin D, Zhu Y, Lei L, Gong H, Mei Y, Teo HY, Wu D, Liu H. IL-12/IL-18-preactivated donor NK cells enhance GVL effects and mitigate GvHD after allogeneic hematopoietic stem cell transplantation. Eur J Immunol 2018; 48:670-682. [PMID: 29282719 DOI: 10.1002/eji.201747177] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 11/25/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022]
Abstract
Adoptive transfer of donor NK cells has the potential of mediating graft-versus-leukemia (GVL) effect while suppressing acute graft-versus-host-disease (aGVHD) during allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, these beneficial effects are limited by the transient function of adoptively transferred NK cells. Previous studies demonstrate that cytokine-induced memory-like NK cells that are preactivated by IL-12, IL-15, and IL-18 have enhanced effector functions and long life span in vivo. Here, we investigated the effects of IL-12/18-preactivated and IL-12/15/18-preactivated donor NK cells on GVL and aGVHD in a murine model of allo-HSCT. We found that both IL-12/18- and IL-12/15/18-preactivated NK cells mediated stronger GVL effect than control NK cells mainly due to their elevated activation/cytotoxicity and sustained proliferative potential. Interestingly, we observed that although both IL-12/18- and IL-12/15/18-preactivated NK cells significantly inhibited severe aGVHD, only the IL-12/18-preactivated NK cells maintained the beneficial effect of donor NK cells on mild aGVHD. The IL-12/15/18-preactivated NK cell infusion accelerated aGVHD in the fully-mismatched mild aGVHD model. Our results demonstrated that IL-12/18-preactivated NK cells displayed sustained and enhanced GVL functions, and could mitigate aGVHD despite the severity of the disease. IL-12/18-preactivated donor NK cell infusion may be an effective and safe adoptive therapy after allo-HSCT.
Collapse
Affiliation(s)
- Yuan Song
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bo Hu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yonghao Liu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziqi Jin
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yinsheng Zhang
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dandan Lin
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Zhu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Lei
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huanle Gong
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Mei
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Huey Yee Teo
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Depei Wu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
29
|
IL-35 expression in hepatocellular carcinoma cells is associated with tumor progression. Oncotarget 2018; 7:45678-45686. [PMID: 27329841 PMCID: PMC5216752 DOI: 10.18632/oncotarget.10141] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/03/2016] [Indexed: 01/02/2023] Open
Abstract
IL-35 has recently been demonstrated to play significant roles in the progression of various malignant tumors. We investigated the expression of IL-35 in hepatocellular carcinoma (HCC) and the regulatory mechanisms in HCC progression. Tissue microarray from 75 HCC patients revealed that IL-35 was primarily localized in the cytoplasm of cancer cells and peri-tumoral hepatocytes. Quantitative analysis showed that IL-35 expression was significantly lower in patients in the advanced stages than in the early stages. Significantly lower expression of IL-35 was also observed in HCC patients with higher histological grades, larger tumor size, positive microvascular invasion and lymph node/distant metastasis. IL-35 over-expression in HepG2 cells significantly upregulated HLA-ABC and CD95, reduced activities of MMP-2 and MMP-9, and decreased cell migration, invasion and colony formation capacities. Our data indicated that decreased expression of IL-35 in tumor tissues might contribute to the progression of HCC, and IL-35 may serve as a new therapeutic target for HCC.
Collapse
|
30
|
Zongyi Y, Funian Z, Hao L, Xin W, Ying C, Jialin Z, Yongfeng L, Baifeng L. Interleukin-35 mitigates the function of murine transplanted islet cells via regulation of Treg/Th17 ratio. PLoS One 2017; 12:e0189617. [PMID: 29236782 PMCID: PMC5728515 DOI: 10.1371/journal.pone.0189617] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 11/29/2017] [Indexed: 12/19/2022] Open
Abstract
Pancreatic islet transplantation is a promising treatment for type 1 diabetes (T1D). Interleukin-35 (IL-35) is a recently discovered cytokine that exhibits potent immunosuppressive functions. However, the role of IL-35 in islet transplant rejection remains to be elucidated. In this study, we isolated islet cells of BALB/c mouse and purified CD4+ T cell subsets of a C57BL/6 mouse. The model for islet transplantation was established in vitro by co-culture of the islet cells and CD4+ T cells. IL-35 (20 ng/ml) was administered every other day. Following co-culture, the islet function and Treg/Th17 ratio were analyzed on days 1, 3, and 5. Furthermore, the Th17/Treg ratio was modulated (1:0–2), and the function of islet cells as well as proliferation of Th17 cells were analyzed. T cell sorting was performed using the magnetic bead sorting method; Treg and Th17 count using flow cytometry; cell proliferation detection using the carboxyfluorescein diacetate succinimidyl ester (CFSE) method, and islet function test using the sugar stimulation test. Results showed that Th17 counts increased in the co-culture system. However, after administration of IL-35, the number of Treg cells increased significantly compared to that in the control group (50.7% of total CD4+ T cells on day 5 in IL-35 group vs. 9.5% in control group) whereas the proliferation rate of Th17 cells was significantly inhibited (0.3% in IL-35 group vs. 7.2% in control group on day 5). Reducing the Th17/Treg ratio significantly improved the function of transplanted islets. Treg inhibited Th17 proliferation and IL-35 enhanced this inhibitory effect. IL-35 mitigates the function of murine transplanted islet cells via regulation of the Treg/Th17 ratio. This might serve as a potential therapeutic strategy for in-vivo islet transplant rejection and T1D.
Collapse
Affiliation(s)
- Yin Zongyi
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen, China
| | - Zou Funian
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Li Hao
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Wang Xin
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Cheng Ying
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China
- Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| | - Zhang Jialin
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China
- Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| | - Liu Yongfeng
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China
- Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| | - Li Baifeng
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
- National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China
- Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
- * E-mail:
| |
Collapse
|
31
|
Han J, Ma S, Gong H, Liu S, Lei L, Hu B, Xu Y, Liu H, Wu D. Inhibition of Acute Graft-versus-Host Disease with Retention of Graft-versus-Tumor Effects by Dimethyl Fumarate. Front Immunol 2017; 8:1605. [PMID: 29209333 PMCID: PMC5702003 DOI: 10.3389/fimmu.2017.01605] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) remains a clinical challenge and a major source of morbidity and mortality following allogeneic hematopoietic stem cell transplantation (allo-HSCT). Dimethyl fumarate (DMF), an activator of Nrf2, has been shown to have anti-inflammatory and immunomodulatory properties without significant immunosuppression. We therefore hypothesized that DMF could be potentially harnessed for the treatment of aGVHD with retention of graft-versus-tumor effect. In this study, we showed that DMF significantly inhibited alloreactive T cell responses in vitro in mixed lymphocyte reaction assay. Administration of DMF significantly alleviated the severity, histological damage, and the overall mortality of aGVHD in an MHC-mismatched aGVHD model. DMF administration reduced the activation and effector function of donor T cells in vitro and in vivo. In addition, DMF treatment upregulated antioxidant enzymes heme oxygenase-1 and glutathione S-transferase-α1 expressions. Furthermore, DMF treatment markedly increased the frequencies of Treg cells. Depletion of CD25+ cells in DMF recipients aggravated aGVHD mortality compared with IgG control recipients. DMF could promote Treg cell differentiation in a dose dependent manner by upregulating TGF-β expression in vitro. Most importantly, DMF administration preserved graft-versus-leukemia effect after bone marrow transplantation. In conclusion, our findings demonstrated DMF as a promising agent for the prevention of aGVHD after allo-HSCT.
Collapse
Affiliation(s)
- Jingjing Han
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Shoubao Ma
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Huanle Gong
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Shuangzhu Liu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Lei
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Bo Hu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yang Xu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haiyan Liu
- Immunology Programme, Department of Microbiology and Immunology, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Depei Wu
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
32
|
Zhou C, Zhang J, Chen Y, Wang H, Hou J. Interleukin-35 as a predictor of prostate cancer in patients undergoing initial prostate biopsy. Onco Targets Ther 2017; 10:3485-3491. [PMID: 28761357 PMCID: PMC5522820 DOI: 10.2147/ott.s135873] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Interleukin (IL)-35 is a novel inhibitory cytokine and has recently been implicated in tumor immunity. However, the role of IL-35 in prostate cancer (PCa) has not been elucidated. Objective To evaluate the role of plasma IL-35 in the diagnosis and prognosis of PCa in Chinese patients undergoing initial prostate biopsy. Materials and methods Using ELISA, plasma IL-35 levels were measured in 180 patients, who underwent a prostate biopsy. The clinical correlation of IL-35 with clinicopathological parameters was also evaluated. Univariate and multivariate logistic regression and receiver operating characteristic (ROC) curve analysis were performed to establish the role of IL-35 as a clinical biomarker. Results Seventy-five (41.6%) of patients were histopathologically confirmed to have PCa. Plasma IL-35 levels were significantly higher in PCa patients (134.48±78.48 pg/mL) compared to non-PCa patients (67.22±24.08 pg/mL). ROC analysis showed that IL-35 was an independent predictor of PCa. Furthermore, IL-35 was found to be a significantly independent predictor of PCa in a group of patients with prostate-specific antigen levels between 4 and 10 ng/mL; was also able to predict advanced PCa from localized PCa and bone metastasis positive PCa from negative PCa. Conclusion Our data suggest for the first time that plasma IL-35 levels are correlated with PCa and is the independent predictor of PCa progression and metastasis. Thus, IL-35 could be utilized as a potential biomarker for diagnosis and prognosis of PCa, could also aid in decision making and predict the stage of the disease.
Collapse
Affiliation(s)
- Chenchao Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ye Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Hao Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
33
|
Gam R, Shah P, Crossland RE, Norden J, Dickinson AM, Dressel R. Genetic Association of Hematopoietic Stem Cell Transplantation Outcome beyond Histocompatibility Genes. Front Immunol 2017; 8:380. [PMID: 28421078 PMCID: PMC5377073 DOI: 10.3389/fimmu.2017.00380] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 03/16/2017] [Indexed: 12/18/2022] Open
Abstract
The outcome of hematopoietic stem cell transplantation (HSCT) is controlled by genetic factors among which the leukocyte antigen human leukocyte antigen (HLA) matching is most important. In addition, minor histocompatibility antigens and non-HLA gene polymorphisms in genes controlling immune responses are known to contribute to the risks associated with HSCT. Besides single-nucleotide polymorphisms (SNPs) in protein coding genes, SNPs in regulatory elements such as microRNAs (miRNAs) contribute to these genetic risks. However, genetic risks require for their realization the expression of the respective gene or miRNA. Thus, gene and miRNA expression studies may help to identify genes and SNPs that indeed affect the outcome of HSCT. In this review, we summarize gene expression profiling studies that were performed in recent years in both patients and animal models to identify genes regulated during HSCT. We discuss SNP–mRNA–miRNA regulatory networks and their contribution to the risks associated with HSCT in specific examples, including forkheadbox protein 3 and regulatory T cells, the role of the miR-155 and miR-146a regulatory network for graft-versus-host disease, and the function of MICA and its receptor NKG2D for the outcome of HSCT. These examples demonstrate how SNPs affect expression or function of proteins that modulate the alloimmune response and influence the outcome of HSCT. Specific miRNAs targeting these genes and directly affecting expression of mRNAs are identified. It might be valuable in the future to determine SNPs and to analyze miRNA and mRNA expression in parallel in cohorts of HSCT patients to further elucidate genetic risks of HSCT.
Collapse
Affiliation(s)
- Rihab Gam
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Centre Göttingen, Göttingen, Germany
| | - Rachel E Crossland
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Jean Norden
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Anne M Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Centre Göttingen, Göttingen, Germany
| |
Collapse
|
34
|
Pylayeva-Gupta Y. Molecular Pathways: Interleukin-35 in Autoimmunity and Cancer. Clin Cancer Res 2016; 22:4973-4978. [PMID: 27582486 DOI: 10.1158/1078-0432.ccr-16-0743] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/12/2016] [Indexed: 11/16/2022]
Abstract
Immunosuppressive functions conferred by regulatory cytokines are important for maintaining homeostasis in immune responses. IL35 has recently emerged as a novel regulator of immune responses. Once thought to be specifically expressed by T regulatory cells, induction of IL35 expression has now been detected in multiple cell types in a variety of diseases, prompting research into regulation of its expression, signaling specificity, target cell populations, and functional outputs. Recent studies have revealed that by directing de novo generation of regulatory T and B cells and inhibiting T effector responses, IL35 plays an important role in the development of autoimmune diseases and cancer. IL35 is overexpressed in a variety of cancers and may exert its function both on antitumor immune responses as well as directly on tumor cells. As such, IL35 is rapidly emerging as a promising biomarker and an attractive cancer therapy target. Clin Cancer Res; 22(20); 4973-8. ©2016 AACR.
Collapse
Affiliation(s)
- Yuliya Pylayeva-Gupta
- Department of Genetics, The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
35
|
Blockade of interleukin-27 signaling reduces GVHD in mice by augmenting Treg reconstitution and stabilizing Foxp3 expression. Blood 2016; 128:2068-2082. [PMID: 27488350 DOI: 10.1182/blood-2016-02-698241] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/25/2016] [Indexed: 02/07/2023] Open
Abstract
Reestablishment of competent regulatory pathways has emerged as a strategy to reduce the severity of graft-versus-host disease (GVHD), and recalibrate the effector and regulatory arms of the immune system. However, clinically feasible, cost-effective strategies that do not require extensive ex vivo cellular manipulation have remained elusive. In the current study, we demonstrate that inhibition of the interleukin-27p28 (IL-27p28) signaling pathway through antibody blockade or genetic ablation prevented lethal GVHD in multiple murine transplant models. Moreover, protection from GVHD was attributable to augmented global reconstitution of CD4+ natural regulatory T cells (nTregs), CD4+ induced Tregs (iTregs), and CD8+ iTregs, and was more potent than temporally concordant blockade of IL-6 signaling. Inhibition of IL-27p28 also enhanced the suppressive capacity of adoptively transferred CD4+ nTregs by increasing the stability of Foxp3 expression. Notably, blockade of IL-27p28 signaling reduced T-cell-derived-IL-10 production in conventional T cells; however, there was no corresponding effect in CD4+ or CD8+ Tregs, indicating that IL-27 inhibition had differential effects on IL-10 production and preserved a mechanistic pathway by which Tregs are known to suppress GVHD. Targeting of IL-27 therefore represents a novel strategy for the in vivo expansion of Tregs and subsequent prevention of GVHD without the requirement for ex vivo cellular manipulation, and provides additional support for the critical proinflammatory role that members of the IL-6 and IL-12 cytokine families play in GVHD biology.
Collapse
|
36
|
Kong B, Liu GB, Zhang JA, Fu XX, Xiang WY, Gao YC, Lu YB, Wu XJ, Qiu F, Wang WD, Yi LL, Zhong JX, Chen ZW, Xu JF. Elevated serum IL-35 and increased expression of IL-35-p35 or -EBI3 in CD4(+)CD25(+) T cells in patients with active tuberculosis. Am J Transl Res 2016; 8:623-633. [PMID: 27158354 PMCID: PMC4846911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/31/2015] [Indexed: 06/05/2023]
Abstract
Despite the recent appreciation of interleukin 35 (IL-35) function in inflammatory diseases, little is known for IL-35 response in patients with active tuberculosis (ATB). In the current study, we demonstrated that ATB patients exhibited increases in serum IL-35 and in mRNA expression of both subunits of IL-35 (p35 and EBI3) in white blood cells and peripheral blood mononuclear cells. Consistently, anti-TB drug treatment led to reduction in serum IL-35 level and p35 or EBI3 expression. TB infection was associated with expression of p35 or EBI3 protein in CD4(+) but not CD8(+) T cells. Most p35(+)CD4(+) T cells and EBI3(+)CD4(+) T cells expressed Treg-associated marker CD25. Our findings may be important in understanding immune pathogenesis of TB. IL-35 in the blood may potentially serve as a biomarker for immune status and prognosis in TB.
Collapse
Affiliation(s)
- Bin Kong
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsNo. 1 Xincheng Road, Dongguan 523808, China
| | - Gan-Bin Liu
- Dongguan 6 People’s HospitalDongguan 523008, China
| | - Jun-Ai Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsNo. 1 Xincheng Road, Dongguan 523808, China
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, China
| | - Xiao-Xia Fu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsNo. 1 Xincheng Road, Dongguan 523808, China
| | - Wen-Yu Xiang
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, China
| | - Yu-Chi Gao
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, China
| | - Yuan-Bin Lu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsNo. 1 Xincheng Road, Dongguan 523808, China
| | - Xian-Jing Wu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsNo. 1 Xincheng Road, Dongguan 523808, China
- Department of Clinical Laboratory, Affiliated Hospital of Guangdong Medical collegeZhanjiang 524001, China
| | - Feng Qiu
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Traditional Chinese MedicineGuangzhou 510120, China
| | - Wan-Dang Wang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsNo. 1 Xincheng Road, Dongguan 523808, China
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of MedicineChicago 60612, Illinois, United States of America
| | - Lai-Long Yi
- Dongguan 6 People’s HospitalDongguan 523008, China
| | - Ji-Xin Zhong
- Department of Medicine, University of Maryland School of MedicineBaltimore, Maryland 21201, United States of America
| | - Zheng W Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of MedicineChicago 60612, Illinois, United States of America
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsNo. 1 Xincheng Road, Dongguan 523808, China
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, China
| |
Collapse
|
37
|
Jin L, Xu X, Ye B, Pan M, Shi Z, Hu Y. Elevated serum interleukin-35 levels correlate with poor prognosis in patients with clear cell renal cell carcinoma. Int J Clin Exp Med 2015; 8:18861-18866. [PMID: 26770508 PMCID: PMC4694408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/05/2015] [Indexed: 06/05/2023]
Abstract
Increasing evidences have demonstrated that serum interleukin-35 (IL-35) levels are closely associated with the development, progression, and poor prognosis of a variety of cancers. However, the relationship between IL-35 and the progression of human clear cell renal cell carcinoma (ccRCC) are poorly understood. The aim of present study was to assess the expression of IL-35 and determine its clinical significance in human ccRCCs. Enzyme-linked immunosorbent assay (ELISA) was performed to examine the serum IL-35 levels in 132 patients with ccRCC and 100 healthy controls. The association of IL-35 levels with clinicopathological parameters and prognosis of ccRCC patients was statistically analyzed. Serum IL-35 levels in patients with ccRCC (25.86±11.78 pg/ml) were significantly higher than those in healthy controls (10.05±9.47 pg/ml, P<0.001). High serum IL-35 levels were significantly correlated with pathologic stage (P<0.001), fuhrman grade (P<0.001), tumor size (P=0.012), T stage (P=0.007), N classification (P=0.002), metastasis (P<0.001) and recurrence (P=0.001). The Kaplan-Meier survival analysis demonstrated that high serum IL-35 levels were significantly associated with poor overall survival (log-rank, P<0.001). Multivariate analysis demonstrated that serum IL-35 levels (HR=2.919, 95% CI =1.871-4.830, P=0.001) and pathologic stage (HR=2.541, 95% CI =1.227-3.987, P=0.002) were an independent prognostic factor for the overall survival of ccRCC patients. In conclusion, high serum IL-35 levels are associated with poor prognosis in patients with ccRCC. IL-35 may represent a promising and useful prognostic biomarker for ccRCC.
Collapse
Affiliation(s)
- Lingwei Jin
- Department of Nephrology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou 325027, Zhejiang, China
| | - Xiaoyan Xu
- Department of Nephrology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou 325027, Zhejiang, China
| | - Bairu Ye
- Department of Nephrology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou 325027, Zhejiang, China
| | - Min Pan
- Department of Nephrology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou 325027, Zhejiang, China
| | - Zhen Shi
- Department of Nephrology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou 325027, Zhejiang, China
| | - Yunling Hu
- Department of Nephrology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou 325027, Zhejiang, China
| |
Collapse
|
38
|
Wang J, Tao Q, Wang H, Wang Z, Wu F, Pan Y, Tao L, Xiong S, Wang Y, Zhai Z. Elevated IL-35 in bone marrow of the patients with acute myeloid leukemia. Hum Immunol 2015; 76:681-6. [DOI: 10.1016/j.humimm.2015.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/16/2015] [Accepted: 09/27/2015] [Indexed: 12/13/2022]
|
39
|
Interleukin-35 administration counteracts established murine type 1 diabetes--possible involvement of regulatory T cells. Sci Rep 2015. [PMID: 26224624 PMCID: PMC4519737 DOI: 10.1038/srep12633] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The anti-inflammatory cytokine IL-35 is produced by regulatory T (Treg) cells to suppress autoimmune and inflammatory responses. The role of IL-35 in type 1 diabetes (T1D) remains to be answered. To elucidate this, we investigated the kinetics of Treg cell response in the multiple low dose streptozotocin induced (MLDSTZ) T1D model and measured the levels of IL-35 in human T1D patients. We found that Treg cells were increased in MLDSTZ mice. However, the Treg cells showed a decreased production of anti-inflammatory (IL-10, IL-35, TGF-β) and increased pro-inflammatory (IFN-γ, IL-2, IL-17) cytokines, indicating a phenotypic shift of Treg cells under T1D condition. IL-35 administration effectively both prevented development of, and counteracted established MLDSTZ T1D, seemingly by induction of Eos expression and IL-35 production in Treg cells, thus reversing the phenotypic shift of the Treg cells. IL-35 administration reversed established hyperglycemia in NOD mouse model of T1D. Moreover, circulating IL-35 levels were decreased in human T1D patients compared to healthy controls. These findings suggest that insufficient IL-35 levels play a pivotal role in the development of T1D and that treatment with IL-35 should be investigated in treatment of T1D and other autoimmune diseases.
Collapse
|
40
|
Sawant DV, Hamilton K, Vignali DAA. Interleukin-35: Expanding Its Job Profile. J Interferon Cytokine Res 2015; 35:499-512. [PMID: 25919641 DOI: 10.1089/jir.2015.0015] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Counter-regulation afforded by specialized regulatory cell populations and immunosuppressive cytokines is critical for balancing immune outcome. The inhibitory potential of the established suppressive cytokines, IL-10 and TGFβ, has been well elucidated in diverse inflammatory scenarios in conjunction with their key roles in Treg development and function. Despite the early predictions for an immunomodulatory role for the Ebi3/p35 heterodimer in placental trophoblasts, IL-35 biology remained elusive until 2007 when it was established as a Treg-restricted inhibitory cytokine. Since then, Treg-derived IL-35 has been shown to exhibit its suppressive activities in a range of autoimmune diseases and cancer models. Recent studies are beginning to explore other cellular sources of IL-35, such as Bregs and CD8(+) Tregs. Despite these new cellular sources and targets, the mode of IL-35 suppression remains restricted to inhibition of proliferation and induction of an IL-35-producing induced regulatory T cell population referred to as iTr35. In this review, we explore the early beginnings, status quo, and future prospects of IL-35 biology. The unparalleled opportunity of targeting multiple immunosuppressive populations (Tregs, Bregs, CD8(+) Tregs) through IL-35 is highly exciting and offers tremendous promise from a translational standpoint, particularly for cancer immunotherapies.
Collapse
Affiliation(s)
- Deepali V Sawant
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Kristia Hamilton
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|